Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Pediatric Nephrology

https://doi.org/10.1007/s00467-018-4093-1

REVIEW

Treatment of steroid-resistant nephrotic syndrome in the genomic era


Adam R. Bensimhon 1 & Anna E. Williams 1 & Rasheed A. Gbadegesin 1,2,3

Received: 6 July 2018 / Revised: 13 September 2018 / Accepted: 18 September 2018


# IPNA 2018

Abstract
The pathogenesis of steroid-resistant nephrotic syndrome (SRNS) is not completely known. Recent advances in genomics have
elucidated some of the molecular mechanisms and pathophysiology of the disease. More than 50 monogenic causes of SRNS
have been identified; however, these genes are responsible for only a small fraction of SRNS in outbred populations. There are
currently no guidelines for genetic testing in SRNS, but evidence from the literature suggests that testing should be guided by the
genetic architecture of the disease in the population. Notably, most genetic forms of SRNS do not respond to current immuno-
suppressive therapies; however, a small subset of patients with monogenic SRNS will achieve partial or complete remission with
specific immunomodulatory agents, presumably due to non-immunosuppressive effects of these agents. We suggest a pragmatic
approach to the therapy of genetic SRNS, as there is no evidence-based algorithm for the management of the disease.

Keywords Nephrotic syndrome . SRNS . Genetic SRNS . Treatment . Genetic SRNS

Introduction children, accounting for about 80% of cases, while the


SRNS variant accounts for the remaining 20% of cases and
Nephrotic syndrome (NS) is the most common glomerular typically portends an unfavorable prognosis [5]. Most chil-
disorder in childhood, with an incidence that ranges from ap- dren with SRNS will progress to end-stage kidney disease
proximately 2 to 7/100,000 children per year and a global (ESKD) within 5–10 years of diagnosis [6, 7].
prevalence of approximately 16/100,000 children [1, 2]. It is The etiology of SRNS is unknown in the majority of chil-
diagnosed by a constellation of clinical signs and symptoms dren with the disease; however, data from genomic studies in
that include massive proteinuria, hypoalbuminemia, edema, the last 20 years suggest that most cases of SRNS are probably
and hyperlipidemia. Childhood nephrotic syndrome is classi- due to structural and functional defects of the podocyte, the
fied based on initial response to a standardized corticosteroid glomerular visceral epithelial cell. Strikingly, the majority of
therapy into steroid-sensitive nephrotic syndrome (SSNS) or the over 50 single-gene causes of hereditary NS are critical for
steroid-resistant nephrotic syndrome (SRNS). Despite the lack the functional integrity of the podocyte; thus, most clinicians
of a global consensus regarding the definition of steroid resis- now refer to SRNS as a podocytopathy [8].
tance, most experts agree that SRNS constitutes the failure to The prevalence of genetic SRNS is unknown due to the
achieve remission after 4–6 weeks of daily corticosteroid ther- dearth of population-based studies. Reported prevalence
apy [3, 4]. The SSNS variant is the most common among varies between 4 and 30%, with a higher prevalence among
inbred populations and a lower prevalence in heterogeneous
Electronic supplementary material The online version of this article outbred populations [9–13]. Sadowski et al. [9] studied 1783
(https://doi.org/10.1007/s00467-018-4093-1) contains supplementary unrelated families and found the prevalence of monogenic NS
material, which is available to authorized users.
to be 29.5%; however, mutation prevalence varies widely be-
tween different regions of the world, with the highest preva-
* Rasheed A. Gbadegesin
rasheed.gbadegesin@duke.edu lence in regions of the world where consanguinity is common.
In addition, there is currently no data from Africa on disease
1
Department of Pediatrics, Division of Nephrology, Duke University burden due to monogenic NS. While genetic SRNS is being
Medical Center, Durham, NC 27710, USA increasingly recognized, there is no standardized approach to
2
Department of Medicine, Division of Nephrology, Duke University the management of children with genetic NS. In addition, the
Medical Center, Durham, NC 27710, USA relationship between genetic mutations and response to ther-
3
Duke Molecular Physiology Institute, Durham, NC, USA apy has not been established, but it has been reported that the
Pediatr Nephrol

prevalence of resistance to corticosteroid therapy and other FSGS affect the structure and functions of the podocyte and its
immunosuppressive agents is significantly higher in patients slit diaphragm [8, 23, 24]. These observations have given rise
with genetic NS compared to those with non-genetic disease to the concept of FSGS being a podocytopathy. In addition to
[14, 15], though there are isolated reports in the literature structural defects of the podocyte, putative soluble circulating
describing patients with monogenic SRNS who have factors resulting from immune dysregulation may also cause
responded to immunosuppressive therapies. To further com- podocyte injury in patients with SRNS. Furthermore, maladap-
pound the dilemma of treatment decision in genetic NS, there tive changes such as obesity, and conditions that result in re-
is accumulating evidence from pre-clinical studies that, in ad- duced nephron number (e.g., congenital or acquired solitary
dition to their immunomodulating effects, the effects of im- kidney, or preterm birth), can cause and perpetuate podocyte
munosuppressive agents such as corticosteroids and calcine- injury [25–33]. It should be noted that soluble urokinase-type
urin inhibitors in NS may be partially due to their stabilizing plasminogen activator receptor (suPAR), CD80, and other
effects on the podocyte F-actin cytoskeleton, suggesting that molecules have been linked to the etiology and pathogenesis
these agents may be useful in the treatment of SRNS due to of SRNS, pattern of therapy response, and risk of recurrence
structural defects of the podocyte [16–19]. following kidney transplantation. However, the genetic basis
In this review, we will address the indications for genetic of how these molecules modulate disease course in NS is un-
testing in SRNS and discuss the implications of a positive and known, and this topic will be the subject of another review.
negative genetic test result on management decisions and
long-term prognosis. We will discuss the current evidence
available for making decisions on whether or not to use im- Genetic SRNS
munomodulatory therapy and/or other supportive agents in
the management of SRNS. We will also suggest a practical Monogenic SRNS
approach to handling this clinical scenario in day-to-day
practice. Monogenic SRNS can be of autosomal recessive (AR), auto-
somal dominant (AD), X-linked, or mitochondrial inheritance
[34]. Typically, recessive inheritance is characterized by early
Etiology and pathogenesis of SRNS onset of disease presentation and high penetrance and is some-
times associated with extra-renal manifestations. In contrast,
SRNS is a clinically heterogeneous condition that is associat- dominant inheritance is associated with later onset of disease
ed with different morphologic changes on kidney biopsy. The presentation and incomplete penetrance. Monogenic causes of
most common pathologic variant associated with SRNS in SRNS are probably responsible for a small proportion of all
children is focal segmental glomerulosclerosis (FSGS). cases of SRNS; however, data from studies of this small frac-
FSGS is a pathologic finding that is characterized by focal tion of patients with SRNS has improved our understanding of
glomerulosclerosis or tuft collapse, segmental hyalinosis, disease pathobiology. The landmark discovery of nephrin
IgM deposits on immunofluorescence staining, and podocyte (NPHS1) as the gene mutated in congenital nephrotic syn-
foot process effacement [20, 21]. Clinically, FSGS is charac- drome by Tryggvason and his colleagues [24] drew attention
terized by rapid progression to ESKD within 5–10 years of to the vital role of genetic and molecular analyses in under-
diagnosis [6, 7]. It is increasingly being recognized that mor- standing the pathogenesis of SRNS.
phological changes in kidney biopsies of patients with FSGS With the completion of the sequencing of the human ge-
are heterogeneous, and that the different morphologic types nome and rapid improvements in high-throughput sequencing
may be associated with different disease courses [22], though technology, the rate of discovery of genes causing monogenic
the detailed description of these variants is beyond the scope SRNS has increased dramatically. Mutations in approximately
of this review. Other less common lesions that may cause 50 genes are now known to cause SRNS [9, 35–40]; however,
SRNS include minimal change disease (MCD) and membra- these genes are responsible for less than 20% of all cases of
nous and membranoproliferative glomerulopathy. SRNS, suggesting that SRNS is genetically heterogeneous
The molecular pathogenesis of FSGS and other SRNSs is and there are still many unidentified genetic causes of the
not fully understood; however, there is evidence that SRNS is disease. The majority of SRNS genes are enriched in the
caused by defects in the glomerular filtration barrier (GFB), the podocyte and the slit diaphragm, as well as other components
charge and size-selective barrier that is made up of specialized of the GFB. A list of genetic causes of SRNS and the locali-
fenestrated endothelial cells, the glomerular basement mem- zation of the genes in the GFB is shown in Table 1. Genes
brane (GBM), and glomerular epithelial cells (podocytes). mutated in SRNS are generally rare in the genus population;
Data emanating from studies of familial FSGS suggest that however, data from different cohorts seems to suggest that
the podocyte is the most important component of the GFB in mutations in NPHS1, NPHS2, and PLCE1 are the most com-
that majority of the more than 50 genes mutated in hereditary mon cause of autosomal recessive SRNS, while mutations in
Pediatr Nephrol

Table 1 Genetic causes of


nephrotic syndrome Gene Protein Mode of inheritance

Slit diaphragm genes


NPHS1 Nephrin AR
NPHS2 Podocin AR
PLCE1 Phospholipase C epsilon 1 AR
CD2AP CD2-associated protein AD, AR
TRPC6 Transient receptor potential channel C6 AD
CRB2 Crumbs family member 2 AR
FAT1 FAT atypical cadherin AR
Transcription factors and nuclear genes
WT1 Wilms tumor protein 1 AD
LMX1B LIM homeobox transcription factor 1-beta AD
SMARCL1 SMARCA-like protein AR
NUP93 Nuclear pore complex protein 93 AR
NUP107 Nuclear pore complex protein 107 AR
NUP205 Nuclear pore complex protein 205 AR
XPO5 Exportin 5 AR
E2F3 E2F transcription factor AD
NXF5 Nuclear RNA export factor 5 X-linked recessive
PAX2 Paired box protein 2 AD
LMNA Lamin A and C AD
WDR73 WD repeat domain 73 AR
Cytoskeletal and membrane genes
ACTN4 Alpha-actinin 4 AD
INF2 Inverted formin 2 AD
MYO1E Myosin 1E AR
MAGI2 Membrane-associated guanylate kinase, AR
inverted 2
ANLN Anillin actin binding protein AD
PTPRO Protein tyrosine phosphatase RO AR
EMP2 Epithelial membrane protein 2 AR
CUBN Cubilin AR
PODXL Podocalyxin AD
ARHGAP24 Rho GTPase-activating protein 24 AD
ARHGDIA Rho GDP dissociation inhibitor alpha AR
DLC1 DLC1 Rho GTPase-activating protein AR
KANK 1/2/4 Kidney ankyrin repeat-containing protein AR
ITSN 1/2 Intersectin protein AR
SYNPO Synaptopodin AD
TNS2 Tensin-2 AR
Mitochondrial, lysosomal, metabolic, and cytosolic genes
COQ2 Coenzyme Q2 4-hyroxybenzoate AR
polyprenyl transferase
COQ6 Coenzyme Q6 monooxygenase AR
PDSS2 Prenyl-diphosphate synthase subunit 2 AR
ADCK4 AarF domain-containing kinase 4 AR
SCARB2 Scavenger receptor class B, member 2 AR
PMM2 Phosphomannomutase 2 AR
ALG1 Asparagine-linked glycosylation 1 AR
TTC21B Tetratricopeptide repeat protein 21B AR
CDK20 Cyclin-dependent kinase 20 AR
Pediatr Nephrol

Table 1 (continued)
Gene Protein Mode of inheritance

CFH Complement factor H AR


DGKE Diacylglycerol kinase epsilon AR
SPGL1 Sphingosine-1-phosphate lyase 1 AR
Glomerular basement membrane genes
LAMB2 Laminin subunit beta-2 AR
ITGB4 Integrin beta 4 AR
ITGA3 Integrin alpha 3 AR
COL4A 3/4/5 Type IV collagen alpha 3, 4, 5 AR, AD, X-linked
Endosomal regulator genes
GAPVD1 GTPase-activating protein and VPS9 AR
domain 1
ANKFY1 Ankyrin repeat and FYVE domain AR
containing 1

AR autosomal recessive, AD autosomal dominant

INF2, COL4A3, COL4A4, WT1, TRPC6, ACTN4, and MCD in Japanese adults [55]. It should be noted that variants
LMX1B are responsible for most cases of autosomal dominant in polygenic genes are not causal and they will result in dis-
SRNS [11, 40–50]. It should be noted that the prevalence of ease only in the presence of additional modifier variants and
mutations in these genes also varies widely between different environmental factors. For example, only about 25% of indi-
regions of the world; for example, R138Q and R138X muta- viduals with the high-risk APOL1 genotype will develop CKD
tions in NPHS2 represent founder mutations in Central Europe in their lifetime.
[51]. More recently, Winkler et al. reported that NPHS2
V260E may also represent a founder mutation among blacks
in South Africa (Cheryl Winkler, PhD, personal communica- The role of genetic testing in SRNS
tion, 26 June 2018). Generally, patients with genetic SRNS
progress more rapidly to ESKD than those with non-genetic Genetic testing in clinical practice is emerging as an invalu-
disease [12]; however, the rate of disease recurrence post- able diagnostic and prognostic tool in the management of
transplantation in those with genetic disease is much lower children with SRNS. Mutation detection in patients with
than that in those with non-genetic disease. In some studies, SRNS potentially allows for (1) a pragmatic approach to the
recurrence risk in genetic SRNS was reported as less than 8% use of different immunosuppressive agents and avoidance of
compared with 30% in patient populations with non-genetic side effects [56], (2) selection of targeted therapies that may
causes [12, 14, 15, 52]. induce remission and/or delay progression to ESKD [57], (3)
prediction of clinical course and post-transplant disease recur-
Polygenic SRNS rence [58], and (4) genetic counseling and possible antenatal
screening [59]. In addition to these clinical benefits, the con-
A full discussion of the role of polygenic inheritance in SRNS tinued discovery of novel SRNS genes will further our under-
is beyond the scope of this review; however, it is worth noting standing of the pathogenesis of SRNS, aid in the precise def-
that two variants in APOL1 gene [G1 allele (rs73885319 or inition of disease phenotype, and allow for a personalized
rs60910145) and G2 allele (rs717853136)] were recently approach to therapy.
identified as being responsible for excess risk of FSGS and
chronic kidney disease (CKD) in African Americans [53]. Genetic testing methods
Heterozygosity for either of these alleles protects against
sleeping sickness, thereby conferring a survival advantage With improvements in high-throughput sequencing technolo-
similar to protection provided by sickle cell trait (hemoglobin gy, genetic testing is now widely available and is being de-
AS) against malaria. However, when the G1 and G2 alleles are ployed by clinicians in routine evaluation of children with
inherited in a homozygous or a compound heterozygous man- SRNS. The most common methods used in the clinical setting
ner, these alleles confer a 7- to 17-fold increased odds for are the sequencing of a panel of candidate genes using direct
FSGS and a plethora of other forms of CKD in African sequencing (Sanger sequencing) and targeted sequencing of
Americans [53, 54]. In addition, variants in glypican-5 candidates (TSC) using a next-generation sequencing platform.
(GPC5) were reported to be associated with FSGS and The latter approach is generally more practical, less laborious,
Pediatr Nephrol

and more cost-effective than direct sequencing, considering the group that mutation detection rate in patients with a family
fact that there are over 50 genes known to cause SRNS, and the history of SRNS and/or CKD is about 40% compared with a
number of causal mutations will likely continue to increase. 6% rate of detection in those without a family history of CKD
The advantages of the TSC approach include (1) high yield (Varner et al., manuscript in preparation).
of positive tests, especially in populations where mutation in
a particular gene or set of genes is known to be common; (2) the
ability to establish causality without extensive family studies; Approaches to management of SRNS
and (3) the ability to more easily manage data from this ap-
proach compared with whole exome sequencing (WES) or Specific therapy
whole genome sequencing (WGS). The major limitation of
the TSC approach is that mutations in novel genes cannot be Corticosteroids
identified, and a negative result does not necessarily rule out
genetic SRNS since the individual may have a mutation in a The initial treatment for all children presenting with NS gener-
yet-to-be-discovered novel gene. ally includes daily corticosteroid therapy for 6 weeks followed
The other approach that is less commonly used in clinical by alternate-day therapy for another 6 weeks, for a total of
practice is WES, where enrichment strategies are used to se- 3 months of treatment [4]. Steroid resistance is defined as a
quence only the protein-coding regions of the gene (exome), failure to achieve remission following 6 weeks of daily therapy.
which accounts for only 1% of the entire human genome, or It should be noted that more than 90% of children who are
WGS, where both the coding and the non-coding regions are steroid-sensitive will achieve remission within 4 weeks of ther-
sequenced. The advantages of WES and WGS are that they apy initiation [60]. In contrast, adult literature seems to suggest
are all inclusive and mutations may be detected in both can- that longer duration of therapy may be needed before classify-
didate and novel genes, especially with the availability of ing an adult as being steroid-resistant [61–64]. The mecha-
informative pedigrees. However, compared with TSC, WES nisms of action of corticosteroids in NS are not fully elucidated;
and WGS are more expensive, and they are more likely to however, the premise for their use is based on the evidence that
generate uninterpretable data especially when informative NS is an immunological disease and corticosteroids act by
pedigrees are not available. In addition, this approach may suppressing T lymphocyte-mediated responses [25]. In addi-
identify potential pathogenic variants that are irrelevant to tion to immunomodulatory effects, there are pre-clinical data
SRNS but have potential consequences on the health and to suggest that corticosteroids may have non-immunologic ef-
well-being of the patients. fects on the F-actin cytoskeleton of the podocyte [17]. In a
study to determine the effects of glucocorticoids on podocytes,
Indications for genetic testing Xing et al. [65] showed that dexamethasone upregulates the
expression of nephrin and tubulin-α in cultured human and
While various genetic testing modalities are now widely avail- murine podocytes, suggesting a direct effect of glucocorticoids
able, there are no guidelines for when or on whom genetic on the podocyte actin cytoskeleton and regulation of critical slit
testing should be done. However, if we view genetic testing diaphragm proteins. More recently, Zhao et al. [18] demonstrat-
like any other diagnostic test, clinicians should consider ed that α-actinin 4 (ACTN4) interacts with the glucocorticoid
whether the test result is apt to alter the management of the receptor (GR) in the nucleus of human podocytes.
patient or better inform discussion of disease prognosis and Glucocorticoids have also been shown to protect and enhance
genetic counseling [34]. If testing will provide any of this recovery of cultured podocytes from puromycin
information, genetic testing is probably indicated. aminonucleoside (PAN)-induced injuries via actin filament sta-
Based on the variable prevalence of genetic SRNS in dif- bilization and protection from apoptosis [66, 67]. Recently,
ferent populations, the approach to genetic testing will depend Mallipattu et al. [68] demonstrated that in vitro treatment with
on where the clinician is practicing. For example, in regions of dexamethasone induced a rapid increase in Krüppel-like factor
the world where inbreeding is highly prevalent or in popula- 15 (KLF15) expression in human and murine podocytes.
tions where there is high prevalence of a particular founder KLF15 is a kidney-enriched zinc finger transcription factor that
mutation, one can make a case for routine genetic testing for has been shown to be required for restoring podocyte differen-
all children with SRNS by sequential TSC followed by WES tiation markers in cultured murine and human podocytes sub-
or WGS if no mutation is detected (Fig. 1a). On the other jected to stress [69]. Overall, these data suggest that in addition
hand, in an outbred population where the likelihood of a pos- to the effects of corticosteroids on T cell function, they may also
itive test is low, genetic testing may be limited to children with have direct effects on the podocyte cytoskeleton and slit dia-
onset of disease in infancy, family history of SRNS or CKD, phragm, indicating that corticosteroids may be useful in the
and syndromic SRNS (Fig. 1b) [9, 13, 34]. A strong point in treatment of some forms of genetic NS. There are few reports
support of the latter approach is a recent finding from our in the literature describing children with monogenic NS who
Pediatr Nephrol

Fig. 1 Approach to genetic testing in SRNS. a Approach to genetic testing in population with high prevalence of SRNS such as an inbred population or
populations with founder mutations. b Approach to genetic testing in an outbred population

have achieved complete or partial remission following cortico- comprehensive study to date is the National Institutes of
steroid therapy (Supplementary Table 1) [70–72]. Based on Health (NIH)-sponsored multicenter randomized trial of 192
published data, there is currently no justification for extended children and young adults with steroid-resistant FSGS who
corticosteroid therapy in children with genetic SRNS. received either CsA or a combination of MMF and pulse
dexamethasone. In this study, 65% of patients in the CsA
Second-line immunomodulatory agents arm achieved remission compared with 42% of those in
MMF/dexamethasone arm. Though the difference in remis-
In patients with SRNS, most clinicians will elect to use sion was not significant, this study demonstrated that both
second-line non-glucocorticoid agents to induce complete or treatments are useful in the management of children with
partial remission, as induction of even partial remission can SRNS [77]. In addition to immunosuppressive effects, the
delay disease progression. Commonly used agents include anti-proteinuric effects of CNIs may also be mediated by
calcineurin inhibitors (CNIs), anti-proliferative agents such intra-renal hemodynamic changes and also by inhibition of
as mycophenolate mofetil (MMF), and biologics like anti- calcineurin-mediated degradation of synaptopodin and stabi-
CD20 (rituximab). A brief description of the mechanism of lization of the podocyte actin cytoskeleton [17]. In another
action of these agents and the rationale for their use in SRNS is study, CNIs were shown to stabilize the podocyte actin cyto-
provided below. skeleton by upregulation of cofilin-1 expression [78]. These
studies suggest that, like glucocorticoids, the non-
Calcineurin inhibitors The two CNIs routinely used to treat NS immunomodulatory effects of CNIs may also ameliorate the
are cyclosporine A (CsA) and tacrolimus (also known as clinical manifestations of genetic SRNS that are due to struc-
FK506). The immunosuppressive properties of CsA and ta- tural defects of podocytes and other components of the GFB.
crolimus result from inhibition of calcineurin, a calcium- and Despite these potential effects on the GFB, a small case series
calmodulin-dependent phosphatase [73, 74]. Both agents in- from the literature seemed to suggest that the majority of chil-
hibit calcineurin, CsA by binding to cyclophilin, and FK506 dren with genetic SRNS will not respond to CNI-based ther-
by binding to protein 12 to form the FKBP12 complex, which apy [56, 79, 80]. In a study of 91 children with SRNS, Buscher
ultimately inhibits calcineurin [73, 74]. Inhibition of calcine- et al. [79] reported that 55% of children with non-genetic
urin results in suppression of the transcription of interleukin-2 SRNS achieved complete remission and 13% were in partial
and subsequent T cell activation [75, 76]. A number of ran- remission following treatment with CNIs compared with 0%
domized clinical trials have suggested improved remission complete remission and 29% partial remission in children with
rates following CNI therapy in patients with SRNS. The most genetic SRNS who presented after the age of 3 months. A
Pediatr Nephrol

similar pattern has been reported in other studies [14, 15, 56, of targeted therapeutic agents. Despite the fact that multiple
70, 81–86]. A summary of studies reporting on the use of genetic causes of NS have been identified, there is a signifi-
CNIs and other therapeutic agents in children with genetic cant lag in the utilization of this information to identify
SRNS can be found in Supplementary Table 1. The major targeted therapies for NS [103]. There are multiple reasons
challenge in the use of CNIs and other second-line agents in for this lag, including but not limited to a lack of high-
the treatment of children with genetic SRNS is that there are fidelity cell lines to study podocytes in culture [103]. The
no clinical or genetic predictors of response to therapy, nor is mouse- and human-derived podocyte cell lines that are cur-
there a guideline on when to stop therapy in unresponsive rently used for biochemical characterization of disease-
patients. causing mutations have provided some insight into pathways
that are defective in patients with genetic NS; however, they
Mycophenolate mofetil MMF is an anti-proliferative agent have some major limitations because they lack essential
that is used extensively for immunosuppression in solid organ podocyte characteristics that may be important to achieving
transplantation and has also been increasingly used for treat- a full understanding of the mechanisms of podocyte injury
ment of SRNS [15, 87, 88]. MMF acts through its active me- [103]. For example, these cell lines do not form slit diaphragm
tabolite mycophenolic acid (MPA) as a non-competitive inhib- cross-bridges in vitro, thereby limiting our understanding of
itor of the enzyme inosine monophosphate dehydrogenase the structural and signaling alterations induced by disease-
(IMPDH), which preferentially inhibits B and T lymphocyte causing mutations [103]. In addition, mouse models that are
proliferation. Its mechanism of action in the treatment of glo- often used to study these mutations in vivo do not accurately
merular disease is not fully understood, but it has been shown recapitulate phenotypes seen in humans [103]. Furthermore,
in both human and experimental studies that MMF may act by pathway analyses are not automated, and they are often labo-
suppressing lymphocyte proliferation and antibody produc- rious and frequently require investigators with different skill
tion. MMF also decreases interleukin-2, interleukin-4, and ad- sets to carry out. Despite these limitations, some of these dis-
hesion molecule expression in the kidney [89–92]. Limited coveries have led to identification of targeted therapies, novel
clinical data suggest that MMF may induce complete or partial therapeutic targets, or existing agents that may be repurposed
remission in steroid- and CsA-resistant FSGS without causing for the treatment of SRNS.
the side effects of nephrotoxicity that are seen with CNIs [93,
94]. In the NIH trial described above, a combination of MMF Coenzyme Q10 supplementation
and dexamethasone was found to be as effective as CsA in the
treatment of SRNS [77]. To date, there are no reports in the Coenzyme Q (CoQ) is a small lipophilic molecule that is
literature on the use of MMF in children with genetic SRNS. synthesized ubiquitously in the inner mitochondrial mem-
brane and is involved in many essential cellular processes,
Rituximab Rituximab is a genetically engineered chimeric especially in the mitochondria [104]. The molecule is com-
monoclonal antibody, containing murine variable regions and posed of a quinone group and a polyisoprenoid tail of variable
a human IgG1 constant domain directed against the CD20 an- length, depending on the species [105]. In humans, CoQ with
tigen expressed on the surface of B lymphocytes [95]. There are 10 isoprenoid subunits is found abundantly in the mitochon-
a few case series in the literature to show that rituximab may dria and is named coenzyme Q10 (CoQ10) or ubiquinone
induce partial and complete remission in some children with [106]. Aside from being an essential component of the mito-
SRNS [96–101]. The mechanisms of action of rituximab in chondrial electron transport chain, CoQ10 is required for py-
both SSNS and SRNS are unknown; however, it has been rimidine nucleoside biosynthesis and is one of the most potent
shown that rituximab directly modulates and protects podocytes lipophilic antioxidants [104, 107]. Biosynthesis of CoQ10 re-
from injury by preserving sphingomyelin phosphodiesterase quires at least 16 different genes (PDSS1, PDSS2, COQ2,
acid-like 3b (SMPDL-3b) expression, suggesting that rituximab COQ3, COQ4, COQ5, COQ6, COQ7, COQ8A, COQ8B,
may modulate podocyte function independent of its immuno- COQ9, COQ10A, COQ10B, FDX1L, FDXR, and
modulatory effects [102]. There are currently no reports to show ALDH3A1) [108–110]. Mutations in some of these genes
that rituximab may be useful in the treatment of genetic SRNS. cause primary CoQ deficiency, a severe but treatable, mito-
chondrial cytopathy [111]. In contrast to most mitochondrial
Agents targeting pathways that are dysregulated respiratory chain disorders, for which no effective treatment
by specific mutations exists, children with primary CoQ10 deficiency respond to oral
supplementation [112]. Mutations in COQ6 and COQ2 have
One of the promises of the genomic revolution is that identi- been reported to cause syndromic and isolated SRNS [57, 111,
fication of genetic causes and genetic risk factors for SRNS 113]. Since the products of these genes have been identified as
will lead to a better understanding of disease pathogenesis, being important in the synthesis of CoQ10, there are reports on
identification of novel diagnostic tools, and the development the use of CoQ10 supplementation in the treatment of children
Pediatr Nephrol

with SRNS due to mutation in these genes [114]. Montini et signaling axis and activation of mTOR-driven endoplasmic
al. [112] described two siblings with missense mutations in reticulum (ER) stress are responsible for the aberrant podocyte
COQ2, one of whom was already in ESKD at presentation and phenotype seen in cell lines overexpressing mutant ANLN
the other sibling presented with NS and was treated with oral [122]. Furthermore, we identified existing compounds and
CoQ10 supplementation. This individual achieved partial re- novel compounds that may reverse the abnormal podocyte
mission and has stable renal function after 5 years of follow- phenotype, showing that rational therapeutic targets for famil-
up [112]. Other reports have found the same pattern; however, ial FSGS can be identified through rigorous biochemical char-
treatment is only effective prior to the development of ESKD acterization of dysregulated podocyte phenotypes [122].
[57, 113, 115]. In another study, Heeringa et al. [57] reported
that up to 50% of children with COQ6 mutations achieved
complete or partial remission with CoQ10 supplementation Supportive therapy
with or without an angiotensin-converting enzyme inhibitor
(ACEi). More recently, mutations in the aarF domain- There are multiple publications to show that supportive mea-
containing kinase 4 (ADCK4) gene were reported as a cause sures such as the use of angiotensin receptor blockade to re-
of SRNS [116]. ADCK4 interacts with components of the duce proteinuria, aggressive blood pressure control, and the
CoQ10 biosynthesis pathway, and patients with ADCK4 mu- use of lipid-lowering agents are useful in both non-genetic and
tations have been reported to have reduced cellular CoQ10 genetic SRNS to slow progression of disease [123, 124].
content [116]. In one study, a child with a homozygous frame-
shift mutation in ADCK4 and SRNS was successfully treated
with CoQ10 supplementation [116]. These studies clearly il- Kidney transplantation
lustrate the power of genomics in the identification of thera-
pies based on the underlying mechanisms of disease. Monogenic SRNS portends a highly unfavorable long-term
prognosis, as ~ 50% of patients will develop ESKD requiring
Vitamin B12 dialysis and kidney transplantation within 10 years of diagno-
sis [15]. Fortunately, several studies have shown that the risk
Mutations in the cubilin (CUBN) gene have been reported in of disease recurrence post-transplantation in patients with ge-
children with intermittent proteinuria [117]. Cubilin is a netic SRNS is low compared with those with non-genetic
coreceptor for the intestinal vitamin B12-intrinsic factor com- SRNS [125, 126]. Ding et al. [52] reported a recurrence rate
plex, and it is also essential for tubular reabsorption of protein of 0% (0/25) in genetic SRNS compared with 30% (26/86) in
in the proximal tubule [118]. It is therefore conceivable that those with non-genetic SRNS. We reported the same pattern in
treatment with vitamin B12 may ameliorate proteinuria associ- a large cohort of North American children with ESKD due to
ated with CUBN mutation. However, there are no reports in the FSGS who received kidney transplantation over 10 years
literature demonstrating the benefit of vitamin B12 supplemen- [127]. Because the risk of post-transplant recurrence is very
tation in the treatment of proteinuria due to CUBN mutations. low in genetic SRNS, deceased donor and living-related donor
(LRD) transplant can be planned with great optimism after
Other agents excluding the disease-causing mutations in living donors.
Scrupulous planning, however, will be required prior to
In addition to identification of specific molecules that may be embarking on LRD kidney transplantation from family mem-
replaced to reverse phenotypes induced by genetic SRNS, bers of patients with known single gene defects, regardless of
some SRNS-associated genetic mutations have also been the mode of inheritance [34]. In autosomal recessive condi-
shown to be viable therapeutic targets. For example, mutations tions for example, the effect of reducing renal mass by 50%
in the transient receptor potential cation channel, subfamily C, (i.e., acquired solitary kidney) in a heterozygous carrier is
member 6 (TRPC6) can cause autosomal dominant FSGS unknown, and the risk of developing kidney disease in that
through aberrant calcium ion conductance in podocytes setting as a result of modifier genes and/or environmental
[103, 119]. Therefore, modification of TRPC6 enzymatic ac- factors is also not clear. In autosomal dominant conditions,
tivity may represent a novel therapeutic approach in the treat- family members should not be considered as LRD candidate
ment of FSGS. Indeed, there are already published proof-of- until the mutation has been ruled out in the donor because of
principle data to show that pharmacological targeting of variable penetrance associated with AD gene and the exis-
TRPC6 and other TRPCs is feasible for treating diseases that tence of asymptomatic carriers who may develop kidney dis-
are associated with TRPC overactivity [120, 121]. More re- ease following reduction of kidney mass by 50%, or the po-
cently, in a study designed to identify pathways that are tential of implanting a kidney that may not function optimally
perturbed in FSGS due to mutations in the ANLN gene, we in the recipient [128]. Thus, we do not support the use of LRD
showed that dysregulation of the PI-3K/AKT/mTOR/Rac1 donation from families with known hereditary disease and
Pediatr Nephrol

Fig. 2 Approach to management of a child with non-genetic steroid-resistant nephrotic syndrome (SRNS)

unknown gene defects. If the gene defect is known, LRD predictor of response to most of these agents is the initial
kidneys should only be accepted if the mutation has been ruled response to corticosteroid therapy. As described above,
out in the potential donor. about 60% of patients with SRNS will achieve partial or
complete remission following use of immunomodulatory
agents other than glucocorticoids; however, there are no
Treatment guidelines for non-genetic robust clinical or laboratory predictors of response to these
and genetic SRNS agents. Despite this lack of predictors, there are guidelines
for the therapy of non-genetic SRNS based on accumulated
Children with SRNS are often challenging to manage be- clinical experience and expert opinions; an example is the
cause many of the agents used are often not effective in Kidney Disease Improving Global Outcomes (KDIGO)
majority of patients. In fact, the single most important guideline (Fig. 2) [3]. The overall goal of most of these

Fig. 3 Suggested approach to management of a child with genetic steroid-resistant nephrotic syndrome (SRNS)
Pediatr Nephrol

treatment approaches is to use a combination of immuno- Funding The National Institutes of Health (NIH) and National Institute
of Diabetes and Digestive and Kidney Diseases (NIDDK) grants
modulatory and supportive agents to achieve complete or
5R01DK098135 and 5R01DK094987 to RAG, ARB, and AEW are sup-
partial remission in order to delay progression of disease ported by the Duke Pediatric Research Scholars (DPRS) program.
and development of ESKD. Because overall experience
with treatment of genetic SRNS is limited, there are no Compliance with ethical standards
such treatment guidelines for children with genetic
SRNS. As described in previous sections, most cases of Conflict of interest The authors declare that they have no conflicts of
genetic SRNS are due to defective proteins that are impor- interest.
tant for maintaining the functional and structural integrity
of the podocyte and other components of the slit dia-
phragm. It is therefore logical to assume that most of the References
immunomodulatory agents will not be effective in the
1. Eddy AA, Symons JM (2003) Nephrotic syndrome in childhood.
treatment of genetic SRNS; in fact, most limited small se-
Lancet 362:629–639
ries support this approach, as over 90% of patients with 2. McKinney PA, Feltbower RG, Brocklebank JT, Fitzpatrick MM
genetic SRNS have been reported to be unresponsive to (2001) Time trends and ethnic patterns of childhood nephrotic
different immunomodulatory agents. Thus, the approach syndrome in Yorkshire, UK. Pediatr Nephrol 16:1040–1044
of most clinicians is to offer only supportive therapy and 3. Cattran DC, Feehally J, Cook T, Liu ZH, Fervenza FC, Mezzano
SA, Floege J, Nachman PH, Gipson DS, Praga M, Glassock RJ,
measures to slow progression of CKD in these children. Radhakrishnan J, Hodson EM, Rovin BH, Jha V, Troyanov S, Li
Importantly, there are pre-clinical data and limited case PKT, Wetzels JFM (2012) Kidney disease: improving global out-
series that suggest that immunomodulatory agents may be comes (KDIGO) glomerulonephritis work group. KDIGO clinical
beneficial in the treatment of genetic SRNS, as discussed practice guideline for glomerulonephritis. Kidney Intl Suppl 2:
139–274
in the preceding paragraphs. Therefore, we approach the 4. Ehrich JH, Brodehl J (1993) Long versus standard prednisone
treatment of children with genetic SRNS in a pragmatic therapy for initial treatment of idiopathic nephrotic syndrome in
way by discussing the uncertainty of benefits of therapy children. Arbeitsgemeinschaft fur Padiatrische Nephrologie. Eur J
with parents and also by having a low threshold for Pediatr 152:357–361
5. Children ISoKDi (1981) The primary nephrotic syndrome in chil-
discontinuing these therapies to minimize side effects. dren. Identification of patients with minimal change nephrotic
Figure 3 shows the approach that we typically use in the syndrome from initial response to prednisone. A report of the
treatment of children with genetic SRNS. There is a need International Study of Kidney Disease in Children. J Pediatr 98:
for randomized trials to evaluate the usefulness of existing 561–564
6. Smith JM, Stablein DM, Munoz R, Hebert D, McDonald RA
agents in the treatment of genetic SRNS.
(2007) Contributions of the transplant registry: the 2006 annual
report of the North American Pediatric Renal Trials and
Collaborative Studies (NAPRTCS). Pediatr Transplant 11:366–
373
7. Hildebrandt F (2010) Genetic kidney diseases. Lancet 375:1287–
Conclusions 1295
8. Wiggins RC (2007) The spectrum of podocytopathies: a unifying
Over the past 20 years, advances in genomics have dramat- view of glomerular diseases. Kidney Int 71:1205–1214
ically improved our understanding of the molecular patho- 9. Sadowski CE, Lovric S, Ashraf S, Pabst WL, Gee HY, Kohl S,
genesis of NS. Studies have revealed that genetic SRNS Engelmann S, Vega-Warner V, Fang H, Halbritter J, Somers MJ,
Tan W, Shril S, Fessi I, Lifton RP, Bockenhauer D, El-Desoky S,
cases make up only a small fraction of SRNS in outbred Kari JA, Zenker M, Kemper MJ, Mueller D, Fathy HM, Soliman
populations and that genetic SRNS is heterogeneous. NA, Group SS, Hildebrandt F (2015) A single-gene cause in
Diagnosis of genetic SRNS may allow for a more person- 29.5% of cases of steroid-resistant nephrotic syndrome. J Am
alized approach to therapy and informed discussion of Soc Nephrol 26:1279–1289
10. Lovric S, Fang H, Vega-Warner V, Sadowski CE, Gee HY,
long-term prognosis and post-kidney transplantation out- Halbritter J, Ashraf S, Saisawat P, Soliman NA, Kari JA, Otto
come with families. There are currently no guidelines for EA, Hildebrandt F, Nephrotic Syndrome Study Group (2014)
genetic testing in SRNS; however, based on the published Rapid detection of monogenic causes of childhood-onset ste-
literature, the decision to carry out genetic testing should roid-resistant nephrotic syndrome. Clin J Am Soc Nephrol 9:
1109–1116
be based on the genetic architecture of the disease in the 11. Trautmann A, Bodria M, Ozaltin F, Gheisari A, Melk A, Azocar
population where the clinician is practicing. We suggest a M, Anarat A, Caliskan S, Emma F, Gellermann J, Oh J, Baskin E,
pragmatic approach to the therapy of genetic SRNS, as Ksiazek J, Remuzzi G, Erdogan O, Akman S, Dusek J, Davitaia T,
there is no evidence-based algorithm for the management Ozkaya O, Papachristou F, Firszt-Adamczyk A, Urasinski T, Testa
S, Krmar RT, Hyla-Klekot L, Pasini A, Ozcakar ZB, Sallay P,
of the disease. There is a need for international collabora- Cakar N, Galanti M, Terzic J, Aoun B, Caldas Afonso A,
tive studies to define the genetic architecture of SRNS and Szymanik-Grzelak H, Lipska BS, Schnaidt S, Schaefer F,
approach to its therapy. PodoNet Consortium (2015) Spectrum of steroid-resistant and
Pediatr Nephrol

congenital nephrotic syndrome in children: the PodoNet registry 27. Coward RJ, Foster RR, Patton D, Ni L, Lennon R, Bates DO,
cohort. Clin J Am Soc Nephrol 10:592–600 Harper SJ, Mathieson PW, Saleem MA (2005) Nephrotic plasma
12. Bierzynska A, McCarthy HJ, Soderquest K, Sen ES, Colby E, alters slit diaphragm-dependent signaling and translocates
Ding WY, Nabhan MM, Kerecuk L, Hegde S, Hughes D, Marks nephrin, podocin, and CD2 associated protein in cultured human
S, Feather S, Jones C, Webb NJ, Ognjanovic M, Christian M, podocytes. J Am Soc Nephrol 16:629–637
Gilbert RD, Sinha MD, Lord GM, Simpson M, Koziell AB, 28. Zimmerman SW (1984) Increased urinary protein excretion in the
Welsh GI, Saleem MA (2017) Genomic and clinical profiling of rat produced by serum from a patient with recurrent focal glomer-
a national nephrotic syndrome cohort advocates a precision med- ular sclerosis after renal transplantation. Clin Nephrol 22:32–38
icine approach to disease management. Kidney Int 91:937–947 29. Davin JC (2016) The glomerular permeability factors in idiopathic
13. Sampson MG, Gillies CE, Robertson CC, Crawford B, Vega- nephrotic syndrome. Pediatr Nephrol 31:207–215
Warner V, Otto EA, Kretzler M, Kang HM (2016) Using popula- 30. Weisinger JR, Kempson RL, Eldridge FL, Swenson RS (1974)
tion genetics to interrogate the monogenic nephrotic syndrome The nephrotic syndrome: a complication of massive obesity.
diagnosis in a case cohort. J Am Soc Nephrol 27:1970–1983 Ann Intern Med 81:440–447
14. Buscher AK, Beck BB, Melk A, Hoefele J, Kranz B, 31. Kambham N, Markowitz GS, Valeri AM, Lin J, D’Agati VD
Bamborschke D, Baig S, Lange-Sperandio B, Jungraithmayr T, (2001) Obesity-related glomerulopathy: an emerging epidemic.
Weber LT, Kemper MJ, Tonshoff B, Hoyer PF, Konrad M, Weber Kidney Int 59:1498–1509
S, German Pediatric Nephrology Association (GPN) (2016) 32. Bhathena DB, Julian BA, McMorrow RG, Baehler RW (1985)
Rapid response to cyclosporin A and favorable renal outcome in Focal sclerosis of hypertrophied glomeruli in solitary functioning
nongenetic versus genetic steroid-resistant nephrotic syndrome. kidneys of humans. Am J Kidney Dis 5:226–232
Clin J Am Soc Nephrol 11:245–253 33. Schreuder MF, Langemeijer ME, Bokenkamp A, Delemarre-Van
15. Trautmann A, Schnaidt S, Lipska-Zietkiewicz BS, Bodria M, de Waal HA, Van Wijk JA (2008) Hypertension and
Ozaltin F, Emma F, Anarat A, Melk A, Azocar M, Oh J, Saeed microalbuminuria in children with congenital solitary kidneys. J
B, Gheisari A, Caliskan S, Gellermann J, Higuita LMS, Paediatr Child Health 44:363–368
Jankauskiene A, Drozdz D, Mir S, Balat A, Szczepanska M, 34. Gbadegesin RA, Winn MP, Smoyer WE (2013) Genetic testing in
Paripovic D, Zurowska A, Bogdanovic R, Yilmaz A, Ranchin nephrotic syndrome—challenges and opportunities. Nat Rev
B, Baskin E, Erdogan O, Remuzzi G, Firszt-Adamczyk A, Nephrol 9:179–184
Kuzma-Mroczkowska E, Litwin M, Murer L, Tkaczyk M, 35. Bierzynska A, Soderquest K, Koziell A (2014) Genes and
Jardim H, Wasilewska A, Printza N, Fidan K, Simkova E, podocytes—new insights into mechanisms of podocytopathy.
Borzecka H, Staude H, Hees K, Schaefer F, PodoNet Front Endocrinol (Lausanne) 5:226
Consortium (2017) Long-term outcome of steroid-resistant ne-
36. Ebarasi L, Ashraf S, Bierzynska A, Gee HY, McCarthy HJ, Lovric
phrotic syndrome in children. J Am Soc Nephrol 28:3055–3065
S, Sadowski CE, Pabst W, Vega-Warner V, Fang H, Koziell A,
16. Ding WY, Saleem MA (2012) Current concepts of the podocyte in Simpson MA, Dursun I, Serdaroglu E, Levy S, Saleem MA,
nephrotic syndrome. Kidney Res Clin Pract 31:87–93 Hildebrandt F, Majumdar A (2015) Defects of CRB2 cause
17. Faul C, Donnelly M, Merscher-Gomez S, Chang YH, Franz S, steroid-resistant nephrotic syndrome. Am J Hum Genet 96:153–
Delfgaauw J, Chang JM, Choi HY, Campbell KN, Kim K, 161
Reiser J, Mundel P (2008) The actin cytoskeleton of kidney 37. Miyake N, Tsukaguchi H, Koshimizu E, Shono A, Matsunaga S,
podocytes is a direct target of the antiproteinuric effect of cyclo- Shiina M, Mimura Y, Imamura S, Hirose T, Okudela K, Nozu K,
sporine A. Nat Med 14:931–938 Akioka Y, Hattori M, Yoshikawa N, Kitamura A, Cheong HI,
18. Zhao X, Khurana S, Charkraborty S, Tian Y, Sedor JR, Bruggman Kagami S, Yamashita M, Fujita A, Miyatake S, Tsurusaki Y,
LA, Kao HY (2017) Alpha actinin 4 (ACTN4) regulates gluco- Nakashima M, Saitsu H, Ohashi K, Imamoto N, Ryo A, Ogata
corticoid receptor-mediated transactivation and transrepression in K, Iijima K, Matsumoto N (2015) Biallelic mutations in nuclear
podocytes. J Biol Chem 292:1637–1647 pore complex subunit NUP107 cause early-childhood-onset
19. Gbadegesin RA, Hall G, Adeyemo A, Hanke N, Tossidou I, steroid-resistant nephrotic syndrome. Am J Hum Genet 97:555–
Burchette J, Wu G, Homstad A, Sparks MA, Gomez J, Jiang R, 566
Alonso A, Lavin P, Conlon P, Korstanje R, Stander MC, Shamsan 38. Braun DA, Sadowski CE, Kohl S, Lovric S, Astrinidis SA, Pabst
G, Barua M, Spurney R, Singhal PC, Kopp JB, Haller H, Howell WL, Gee HY, Ashraf S, Lawson JA, Shril S, Airik M, Tan W,
D, Pollak MR, Shaw AS, Schiffer M, Winn MP (2014) Mutations Schapiro D, Rao J, Choi WI, Hermle T, Kemper MJ, Pohl M,
in the gene that encodes the F-actin binding protein anillin cause Ozaltin F, Konrad M, Bogdanovic R, Buscher R, Helmchen U,
FSGS. J Am Soc Nephrol 25:1991–2002 Serdaroglu E, Lifton RP, Antonin W, Hildebrandt F (2016)
20. D’Agati VD, Fogo AB, Bruijn JA, Jennette JC (2004) Pathologic Mutations in nuclear pore genes NUP93, NUP205 and XPO5
classification of focal segmental glomerulosclerosis: a working cause steroid-resistant nephrotic syndrome. Nat Genet 48:457–
proposal. Am J Kidney Dis 43:368–382 465
21. D’Agati VD, Kaskel FJ, Falk RJ (2011) Focal segmental 39. Gee HY, Zhang F, Ashraf S, Kohl S, Sadowski CE, Vega-Warner
glomerulosclerosis. N Engl J Med 365:2398–2411 V, Zhou W, Lovric S, Fang H, Nettleton M, Zhu JY, Hoefele J,
22. Fogo AB (2015) Causes and pathogenesis of focal segmental Weber LT, Podracka L, Boor A, Fehrenbach H, Innis JW,
glomerulosclerosis. Nat Rev Nephrol 11:76–87 Washburn J, Levy S, Lifton RP, Otto EA, Han Z, Hildebrandt F
23. Buscher AK, Weber S (2012) Educational paper: the (2015) KANK deficiency leads to podocyte dysfunction and ne-
podocytopathies. Eur J Pediatr 171:1151–1160 phrotic syndrome. J Clin Invest 125:2375–2384
24. Tryggvason K, Patrakka J, Wartiovaara J (2006) Hereditary pro- 40. McCarthy HJ, Bierzynska A, Wherlock M, Ognjanovic M,
teinuria syndromes and mechanisms of proteinuria. N Engl J Med Kerecuk L, Hegde S, Feather S, Gilbert RD, Krischock L, Jones
354:1387–1401 C, Sinha MD, Webb NJ, Christian M, Williams MM, Marks S,
25. Karp AM, Gbadegesin RA (2017) Genetics of childhood steroid- Koziell A, Welsh GI, Saleem MA, RADAR the UK SRNS Study
sensitive nephrotic syndrome. Pediatr Nephrol 32:1481–1488 Group (2013) Simultaneous sequencing of 24 genes associated
26. Shalhoub RJ (1974) Pathogenesis of lipoid nephrosis: a disorder with steroid-resistant nephrotic syndrome. Clin J Am Soc
of T-cell function. Lancet 2:556–560 Nephrol 8:637–648
Pediatr Nephrol

41. Warejko JK, Tan W, Daga A, Schapiro D, Lawson JA, Shril S, Ettenger R, Antignac C, Wiggins RC, Zenker M, Hildebrandt F
Lovric S, Ashraf S, Rao J, Hermle T, Jobst-Schwan T, Widmeier (2008) Mutations in PLCE1 are a major cause of isolated diffuse
E, Majmundar AJ, Schneider R, Gee HY, Schmidt JM, Vivante A, mesangial sclerosis (IDMS). Nephrol Dial Transplant 23:1291–
van der Ven AT, Ityel H, Chen J, Sadowski CE, Kohl S, Pabst WL, 1297
Nakayama M, Somers MJG, Rodig NM, Daouk G, Baum M, 51. Niaudet P (2004) Podocin and nephrotic syndrome: implications
Stein DR, Ferguson MA, Traum AZ, Soliman NA, Kari JA, El for the clinician. J Am Soc Nephrol 15:832–834
Desoky S, Fathy H, Zenker M, Bakkaloglu SA, Muller D, Noyan 52. Ding WY, Koziell A, McCarthy HJ, Bierzynska A, Bhagavatula
A, Ozaltin F, Cadnapaphornchai MA, Hashmi S, Hopcian J, Kopp MK, Dudley JA, Inward CD, Coward RJ, Tizard J, Reid C,
JB, Benador N, Bockenhauer D, Bogdanovic R, Stajic N, Chernin Antignac C, Boyer O, Saleem MA (2014) Initial steroid sensitivity
G, Ettenger R, Fehrenbach H, Kemper M, Munarriz RL, Podracka in children with steroid-resistant nephrotic syndrome predicts
L, Buscher R, Serdaroglu E, Tasic V, Mane S, Lifton RP, Braun post-transplant recurrence. J Am Soc Nephrol 25:1342–1348
DA, Hildebrandt F (2018) Whole exome sequencing of patients 53. Genovese G, Friedman DJ, Ross MD, Lecordier L, Uzureau P,
with steroid-resistant nephrotic syndrome. Clin J Am Soc Nephrol Freedman BI, Bowden DW, Langefeld CD, Oleksyk TK,
13:53–62 Uscinski Knob AL, Bernhardy AJ, Hicks PJ, Nelson GW,
42. Gbadegesin RA, Lavin PJ, Hall G, Bartkowiak B, Homstad A, Vanhollebeke B, Winkler CA, Kopp JB, Pays E, Pollak MR
Jiang R, Wu G, Byrd A, Lynn K, Wolfish N, Ottati C, Stevens P, (2010) Association of trypanolytic ApoL1 variants with kidney
Howell D, Conlon P, Winn MP (2012) Inverted formin 2 muta- disease in African Americans. Science 329:841–845
tions with variable expression in patients with sporadic and hered- 54. Freedman BI, Kopp JB, Langefeld CD, Genovese G, Friedman
itary focal and segmental glomerulosclerosis. Kidney Int 81:94– DJ, Nelson GW, Winkler CA, Bowden DW, Pollak MR (2010)
99 The apolipoprotein L1 (APOL1) gene and nondiabetic nephropa-
43. Brown EJ, Schlondorff JS, Becker DJ, Tsukaguchi H, Tonna SJ, thy in African Americans. J Am Soc Nephrol 21:1422–1426
Uscinski AL, Higgs HN, Henderson JM, Pollak MR (2010) 55. Okamoto K, Tokunaga K, Doi K, Fujita T, Suzuki H, Katoh T,
Mutations in the formin gene INF2 cause focal segmental Watanabe T, Nishida N, Mabuchi A, Takahashi A, Kubo M,
glomerulosclerosis. Nat Genet 42:72–76 Maeda S, Nakamura Y, Noiri E (2011) Common variation in
44. Barua M, Brown EJ, Charoonratana VT, Genovese G, Sun H, GPC5 is associated with acquired nephrotic syndrome. Nat
Pollak MR (2013) Mutations in the INF2 gene account for a sig- Genet 43:459–463
nificant proportion of familial but not sporadic focal and segmen-
56. Ruf RG, Lichtenberger A, Karle SM, Haas JP, Anacleto FE,
tal glomerulosclerosis. Kidney Int 83:316–322
Schultheiss M, Zalewski I, Imm A, Ruf EM, Mucha B, Bagga
45. Boyer O, Benoit G, Gribouval O, Nevo F, Tete MJ, Dantal J,
A, Neuhaus T, Fuchshuber A, Bakkaloglu A, Hildebrandt F,
Gilbert-Dussardier B, Touchard G, Karras A, Presne C, Grunfeld
Arbeitsgemeinschaft Fur Padiatrische Nephrologie Study Group
JP, Legendre C, Joly D, Rieu P, Mohsin N, Hannedouche T, Moal
(2004) Patients with mutations in NPHS2 (podocin) do not re-
V, Gubler MC, Broutin I, Mollet G, Antignac C (2011) Mutations
spond to standard steroid treatment of nephrotic syndrome. J
in INF2 are a major cause of autosomal dominant focal segmental
Am Soc Nephrol 15:722–732
glomerulosclerosis. J Am Soc Nephrol 22:239–245
57. Heeringa SF, Chernin G, Chaki M, Zhou W, Sloan AJ, Ji Z, Xie
46. Malone AF, Phelan PJ, Hall G, Cetincelik U, Homstad A, Alonso
LX, Salviati L, Hurd TW, Vega-Warner V, Killen PD, Raphael Y,
AS, Jiang R, Lindsey TB, Wu G, Sparks MA, Smith SR, Webb
Ashraf S, Ovunc B, Schoeb DS, McLaughlin HM, Airik R,
NJ, Kalra PA, Adeyemo AA, Shaw AS, Conlon PJ, Jennette JC,
Vlangos CN, Gbadegesin R, Hinkes B, Saisawat P, Trevisson E,
Howell DN, Winn MP, Gbadegesin RA (2014) Rare hereditary
Doimo M, Casarin A, Pertegato V, Giorgi G, Prokisch H, Rotig A,
COL4A3/COL4A4 variants may be mistaken for familial focal
Nurnberg G, Becker C, Wang S, Ozaltin F, Topaloglu R,
segmental glomerulosclerosis. Kidney Int 86:1253–1259
Bakkaloglu A, Bakkaloglu SA, Muller D, Beissert A, Mir S,
47. Lipska BS, Iatropoulos P, Maranta R, Caridi G, Ozaltin F, Anarat
Berdeli A, Varpizen S, Zenker M, Matejas V, Santos-Ocana C,
A, Balat A, Gellermann J, Trautmann A, Erdogan O, Saeed B,
Navas P, Kusakabe T, Kispert A, Akman S, Soliman NA, Krick
Emre S, Bogdanovic R, Azocar M, Balasz-Chmielewska I, Benetti
S, Mundel P, Reiser J, Nurnberg P, Clarke CF, Wiggins RC, Faul
E, Caliskan S, Mir S, Melk A, Ertan P, Baskin E, Jardim H,
Davitaia T, Wasilewska A, Drozdz D, Szczepanska M, C, Hildebrandt F (2011) COQ6 mutations in human patients pro-
Jankauskiene A, Higuita LM, Ardissino G, Ozkaya O, Kuzma- duce nephrotic syndrome with sensorineural deafness. J Clin
Mroczkowska E, Soylemezoglu O, Ranchin B, Medynska A, Invest 121:2013–2024
Tkaczyk M, Peco-Antic A, Akil I, Jarmolinski T, Firszt- 58. Weber S, Gribouval O, Esquivel EL, Moriniere V, Tete MJ,
Adamczyk A, Dusek J, Simonetti GD, Gok F, Gheissari A, Legendre C, Niaudet P, Antignac C (2004) NPHS2 mutation anal-
Emma F, Krmar RT, Fischbach M, Printza N, Simkova E, Mele ysis shows genetic heterogeneity of steroid-resistant nephrotic
C, Ghiggeri GM, Schaefer F, PodoNet Consortium (2013) Genetic syndrome and low post-transplant recurrence. Kidney Int 66:
screening in adolescents with steroid-resistant nephrotic syn- 571–579
drome. Kidney Int 84:206–213 59. Gigante M, Greco P, Defazio V, Lucci M, Margaglione M,
48. Hinkes BG, Mucha B, Vlangos CN, Gbadegesin R, Liu J, Gesualdo L, Iolascon A (2005) Congenital nephrotic syndrome
Hasselbacher K, Hangan D, Ozaltin F, Zenker M, Hildebrandt F, of Finnish type: detection of new nephrin mutations and prenatal
Arbeitsgemeinschaft fur Paediatrische Nephrologie Study Group diagnosis in an Italian family. Prenat Diagn 25:407–410
(2007) Nephrotic syndrome in the first year of life: two thirds of 60. (1981) Primary nephrotic syndrome in children: clinical signifi-
cases are caused by mutations in 4 genes (NPHS1, NPHS2, WT1, cance of histopathologic variants of minimal change and of diffuse
and LAMB2). Pediatrics 119:e907–e919 mesangial hypercellularity. A report of the International Study of
49. Hinkes B, Vlangos C, Heeringa S, Mucha B, Gbadegesin R, Liu J, Kidney Disease in Children. Kidney Int 20:765–771
Hasselbacher K, Ozaltin F, Hildebrandt F, APN Study Group 61. Black DA, Rose G, Brewer DB (1970) Controlled trial of predni-
(2008) Specific podocin mutations correlate with age of onset in sone in adult patients with the nephrotic syndrome. Br Med J 3:
steroid-resistant nephrotic syndrome. J Am Soc Nephrol 19:365– 421–426
371 62. Rydel JJ, Korbet SM, Borok RZ, Schwartz MM (1995) Focal
50. Gbadegesin R, Hinkes BG, Hoskins BE, Vlangos CN, Heeringa segmental glomerular sclerosis in adults: presentation, course,
SF, Liu J, Loirat C, Ozaltin F, Hashmi S, Ulmer F, Cleper R, and response to treatment. Am J Kidney Dis 25:534–542
Pediatr Nephrol

63. Banfi G, Moriggi M, Sabadini E, Fellin G, D’Amico G, Ponticelli 74. Bram RJ, Hung DT, Martin PK, Schreiber SL, Crabtree GR (1993)
C (1991) The impact of prolonged immunosuppression on the Identification of the immunophilins capable of mediating inhibi-
outcome of idiopathic focal-segmental glomerulosclerosis with tion of signal transduction by cyclosporin A and FK506: roles of
nephrotic syndrome in adults. A collaborative retrospective study. calcineurin binding and cellular location. Mol Cell Biol 13:4760–
Clin Nephrol 36:53–59 4769
64. Korbet SM, Schwartz MM, Lewis EJ (1994) Primary focal seg- 75. O’Keefe SJ, Tamura J, Kincaid RL, Tocci MJ, O’Neill EA (1992)
mental glomerulosclerosis: clinical course and response to thera- FK-506- and CsA-sensitive activation of the interleukin-2 promot-
py. Am J Kidney Dis 23:773–783 er by calcineurin. Nature 357:692–694
65. Xing CY, Saleem MA, Coward RJ, Ni L, Witherden IR, 76. Clipstone NA, Crabtree GR (1992) Identification of calcineurin as
Mathieson PW (2006) Direct effects of dexamethasone on human a key signalling enzyme in T-lymphocyte activation. Nature 357:
podocytes. Kidney Int 70:1038–1045 695–697
66. Ransom RF, Lam NG, Hallett MA, Atkinson SJ, Smoyer WE 77. Gipson DS, Trachtman H, Kaskel FJ, Greene TH, Radeva MK,
(2005) Glucocorticoids protect and enhance recovery of cultured Gassman JJ, Moxey-Mims MM, Hogg RJ, Watkins SL, Fine RN,
murine podocytes via actin filament stabilization. Kidney Int 68: Hogan SL, Middleton JP, Vehaskari VM, Flynn PA, Powell LM,
2473–2483 Vento SM, McMahan JL, Siegel N, D’Agati VD, Friedman AL
67. Wada T, Pippin JW, Marshall CB, Griffin SV, Shankland SJ (2011) Clinical trial of focal segmental glomerulosclerosis in chil-
(2005) Dexamethasone prevents podocyte apoptosis induced by dren and young adults. Kidney Int 80:868–878
puromycin aminonucleoside: role of p53 and Bcl-2-related family 78. Li X, Zhang X, Li X, Wang X, Wang S, Ding J (2014)
proteins. J Am Soc Nephrol 16:2615–2625 Cyclosporine A protects podocytes via stabilization of cofilin-1
expression in the unphosphorylated state. Exp Biol Med
68. Mallipattu SK, Guo Y, Revelo MP, Roa-Pena L, Miller T, Ling J,
(Maywood) 239:922–936
Shankland SJ, Bialkowska AB, Ly V, Estrada C, Jain MK, Lu Y,
79. Buscher AK, Kranz B, Buscher R, Hildebrandt F, Dworniczak B,
Ma’ayan A, Mehrotra A, Yacoub R, Nord EP, Woroniecki RP,
Pennekamp P, Kuwertz-Broking E, Wingen AM, John U, Kemper
Yang VW, He JC (2017) Kruppel-like factor 15 mediates
M, Monnens L, Hoyer PF, Weber S, Konrad M (2010)
glucocorticoid-induced restoration of podocyte differentiation
Immunosuppression and renal outcome in congenital and pediatric
markers. J Am Soc Nephrol 28:166–184
steroid-resistant nephrotic syndrome. Clin J Am Soc Nephrol 5:
69. Mallipattu SK, Liu R, Zheng F, Narla G, Ma’ayan A, Dikman S, 2075–2084
Jain MK, Saleem M, D’Agati V, Klotman P, Chuang PY, He JC
80. Giglio S, Provenzano A, Mazzinghi B, Becherucci F, Giunti L,
(2012) Kruppel-like factor 15 (KLF15) is a key regulator of Sansavini G, Ravaglia F, Roperto RM, Farsetti S, Benetti E,
podocyte differentiation. J Biol Chem 287:19122–19135 Rotondi M, Murer L, Lazzeri E, Lasagni L, Materassi M,
70. Hinkes B, Wiggins RC, Gbadegesin R, Vlangos CN, Seelow D, Romagnani P (2015) Heterogeneous genetic alterations in sporad-
Nurnberg G, Garg P, Verma R, Chaib H, Hoskins BE, Ashraf S, ic nephrotic syndrome associate with resistance to immunosup-
Becker C, Hennies HC, Goyal M, Wharram BL, Schachter AD, pression. J Am Soc Nephrol 26:230–236
Mudumana S, Drummond I, Kerjaschki D, Waldherr R, Dietrich 81. Klaassen I, Ozgoren B, Sadowski CE, Moller K, van Husen M,
A, Ozaltin F, Bakkaloglu A, Cleper R, Basel-Vanagaite L, Pohl M, Lehnhardt A, Timmermann K, Freudenberg F, Helmchen U, Oh J,
Griebel M, Tsygin AN, Soylu A, Muller D, Sorli CS, Bunney TD, Kemper MJ (2015) Response to cyclosporine in steroid-resistant
Katan M, Liu J, Attanasio M, O’Toole JF, Hasselbacher K, Mucha nephrotic syndrome: discontinuation is possible. Pediatr Nephrol
B, Otto EA, Airik R, Kispert A, Kelley GG, Smrcka AV, 30:1477–1483
Gudermann T, Holzman LB, Nurnberg P, Hildebrandt F (2006) 82. Stefanidis CJ, Querfeld U (2011) The podocyte as a target: cyclo-
Positional cloning uncovers mutations in PLCE1 responsible for a sporin A in the management of the nephrotic syndrome caused by
nephrotic syndrome variant that may be reversible. Nat Genet 38: WT1 mutations. Eur J Pediatr 170:1377–1383
1397–1405 83. Gellermann J, Stefanidis CJ, Mitsioni A, Querfeld U (2010)
71. Gee HY, Ashraf S, Wan X, Vega-Warner V, Esteve-Rudd J, Lovric Successful treatment of steroid-resistant nephrotic syndrome asso-
S, Fang H, Hurd TW, Sadowski CE, Allen SJ, Otto EA, Korkmaz ciated with WT1 mutations. Pediatr Nephrol 25:1285–1289
E, Washburn J, Levy S, Williams DS, Bakkaloglu SA, 84. Wasilewska AM, Kuroczycka-Saniutycz E, Zoch-Zwierz W
Zolotnitskaya A, Ozaltin F, Zhou W, Hildebrandt F (2014) (2011) Effect of cyclosporin A on proteinuria in the course of
Mutations in EMP2 cause childhood-onset nephrotic syndrome. glomerulopathy associated with WT1 mutations. Eur J Pediatr
Am J Hum Genet 94:884–890 170:389–391
72. Ashraf S, Kudo H, Rao J, Kikuchi A, Widmeier E, Lawson JA, 85. Megremis S, Mitsioni A, Mitsioni AG, Fylaktou I, Kitsiou-Tzelli
Tan W, Hermle T, Warejko JK, Shril S, Airik M, Jobst-Schwan T, S, Stefanidis CJ, Kanavakis E, Traeger-Synodinos J (2009)
Lovric S, Braun DA, Gee HY, Schapiro D, Majmundar AJ, Nucleotide variations in the NPHS2 gene in Greek children with
Sadowski CE, Pabst WL, Daga A, van der Ven AT, Schmidt JM, steroid-resistant nephrotic syndrome. Genet Test Mol Biomarkers
Low BC, Gupta AB, Tripathi BK, Wong J, Campbell K, Metcalfe 13:249–256
K, Schanze D, Niihori T, Kaito H, Nozu K, Tsukaguchi H, Tanaka 86. Malina M, Cinek O, Janda J, Seeman T (2009) Partial remission
R, Hamahira K, Kobayashi Y, Takizawa T, Funayama R, with cyclosporine A in a patient with nephrotic syndrome due to
Nakayama K, Aoki Y, Kumagai N, Iijima K, Fehrenbach H, NPHS2 mutation. Pediatr Nephrol 24:2051–2053
Kari JA, El Desoky S, Jalalah S, Bogdanovic R, Stajic N, 87. Gargah TT, Lakhoua MR (2011) Mycophenolate mofetil in treat-
Zappel H, Rakhmetova A, Wassmer SR, Jungraithmayr T, ment of childhood steroid-resistant nephrotic syndrome. J Nephrol
Strehlau J, Kumar AS, Bagga A, Soliman NA, Mane SM, 24:203–207
Kaufman L, Lowy DR, Jairajpuri MA, Lifton RP, Pei Y, Zenker 88. de Mello VR, Rodrigues MT, Mastrocinque TH, Martins SP, de
M, Kure S, Hildebrandt F (2018) Mutations in six nephrosis genes Andrade OV, Guidoni EB, Scheffer DK, Martini Filho D,
delineate a pathogenic pathway amenable to treatment. Nat Toporovski J, Benini V (2010) Mycophenolate mofetil in children
Commun 9:1960 with steroid/cyclophosphamide-resistant nephrotic syndrome.
73. Flanagan WM, Corthesy B, Bram RJ, Crabtree GR (1991) Pediatr Nephrol 25:453–460
Nuclear association of a T-cell transcription factor blocked by 89. Ziswiler R, Steinmann-Niggli K, Kappeler A, Daniel C, Marti HP
FK-506 and cyclosporin A. Nature 352:803–807 (1998) Mycophenolic acid: a new approach to the therapy of
Pediatr Nephrol

experimental mesangial proliferative glomerulonephritis. J Am 109. Desbats MA, Vetro A, Limongelli I, Lunardi G, Casarin A, Doimo
Soc Nephrol 9:2055–2066 M, Spinazzi M, Angelini C, Cenacchi G, Burlina A, Rodriguez
90. Hauser IA, Renders L, Radeke HH, Sterzel RB, Goppelt-Struebe Hernandez MA, Chiandetti L, Clementi M, Trevisson E, Navas P,
M (1999) Mycophenolate mofetil inhibits rat and human Zuffardi O, Salviati L (2015) Primary coenzyme Q10 deficiency
mesangial cell proliferation by guanosine depletion. Nephrol presenting as fatal neonatal multiorgan failure. Eur J Hum Genet
Dial Transplant 14:58–63 23:1254–1258
91. Penny MJ, Boyd RA, Hall BM (1998) Mycophenolate mofetil 110. Payet LA, Leroux M, Willison JC, Kihara A, Pelosi L, Pierrel F
prevents the induction of active Heymann nephritis: association (2016) Mechanistic details of early steps in coenzyme Q biosyn-
with Th2 cytokine inhibition. J Am Soc Nephrol 9:2272–2282 thesis pathway in yeast. Cell Chem Biol 23:1241–1250
92. Allison AC, Kowalski WJ, Muller CJ, Waters RV, Eugui EM 111. Acosta MJ, Vazquez Fonseca L, Desbats MA, Cerqua C, Zordan
(1993) Mycophenolic acid and brequinar, inhibitors of purine R, Trevisson E, Salviati L (2016) Coenzyme Q biosynthesis in
and pyrimidine synthesis, block the glycosylation of adhesion health and disease. Biochim Biophys Acta 1857:1079–1085
molecules. Transplant Proc 25:67–70 112. Montini G, Malaventura C, Salviati L (2008) Early coenzyme Q10
93. Cattran DC, Wang MM, Appel G, Matalon A, Briggs W (2004) supplementation in primary coenzyme Q10 deficiency. N Engl J
Mycophenolate mofetil in the treatment of focal segmental Med 358:2849–2850
glomerulosclerosis. Clin Nephrol 62:405–411 113. Diomedi-Camassei F, Di Giandomenico S, Santorelli FM, Caridi
94. Montane B, Abitbol C, Chandar J, Strauss J, Zilleruelo G (2003) G, Piemonte F, Montini G, Ghiggeri GM, Murer L, Barisoni L,
Novel therapy of focal glomerulosclerosis with mycophenolate Pastore A, Muda AO, Valente ML, Bertini E, Emma F (2007)
and angiotensin blockade. Pediatr Nephrol 18:772–777 COQ2 nephropathy: a newly described inherited
95. Salama AD, Pusey CD (2006) Drug insight: rituximab in renal mitochondriopathy with primary renal involvement. J Am Soc
disease and transplantation. Nat Clin Pract Nephrol 2:221–230 Nephrol 18:2773–2780
96. Gulati A, Sinha A, Jordan SC, Hari P, Dinda AK, Sharma S, 114. Ozaltin F (2014) Primary coenzyme Q10 (CoQ 10) deficiencies
Srivastava RN, Moudgil A, Bagga A (2010) Efficacy and safety and related nephropathies. Pediatr Nephrol 29:961–969
of treatment with rituximab for difficult steroid-resistant and - 115. Starr MC, Chang IJ, Finn LS, Sun A, Larson AA, Goebel J,
dependent nephrotic syndrome: multicentric report. Clin J Am Hanevold C, Thies J, Van Hove JLK, Hingorani SR, Lam C
Soc Nephrol 5:2207–2212 (2018) COQ2 nephropathy: a treatable cause of nephrotic syn-
97. Prytula A, Iijima K, Kamei K, Geary D, Gottlich E, Majeed A, drome in children. Pediatr Nephrol. https://doi.org/10.1007/
Taylor M, Marks SD, Tuchman S, Camilla R, Ognjanovic M, s00467-018-3937-z
Filler G, Smith G, Tullus K (2010) Rituximab in refractory ne- 116. Ashraf S, Gee HY, Woerner S, Xie LX, Vega-Warner V, Lovric S,
phrotic syndrome. Pediatr Nephrol 25:461–468 Fang H, Song X, Cattran DC, Avila-Casado C, Paterson AD,
98. Ito S, Kamei K, Ogura M, Udagawa T, Fujinaga S, Saito M, Sako Nitschke P, Bole-Feysot C, Cochat P, Esteve-Rudd J,
M, Iijima K (2013) Survey of rituximab treatment for childhood- Haberberger B, Allen SJ, Zhou W, Airik R, Otto EA, Barua M,
onset refractory nephrotic syndrome. Pediatr Nephrol 28:257–264 Al-Hamed MH, Kari JA, Evans J, Bierzynska A, Saleem MA,
99. Bagga A, Sinha A, Moudgil A (2007) Rituximab in patients with Bockenhauer D, Kleta R, El Desoky S, Hacihamdioglu DO, Gok
the steroid-resistant nephrotic syndrome. N Engl J Med 356:2751– F, Washburn J, Wiggins RC, Choi M, Lifton RP, Levy S, Han Z,
2752 Salviati L, Prokisch H, Williams DS, Pollak M, Clarke CF, Pei Y,
100. Magnasco A, Ravani P, Edefonti A, Murer L, Ghio L, Belingheri Antignac C, Hildebrandt F (2013) ADCK4 mutations promote
M, Benetti E, Murtas C, Messina G, Massella L, Porcellini MG, steroid-resistant nephrotic syndrome through CoQ10 biosynthesis
Montagna M, Regazzi M, Scolari F, Ghiggeri GM (2012) disruption. J Clin Invest 123:5179–5189
Rituximab in children with resistant idiopathic nephrotic syn- 117. Ovunc B, Otto EA, Vega-Warner V, Saisawat P, Ashraf S,
drome. J Am Soc Nephrol 23:1117–1124 Ramaswami G, Fathy HM, Schoeb D, Chernin G, Lyons RH,
101. Kamei K, Okada M, Sato M, Fujimaru T, Ogura M, Nakayama M, Yilmaz E, Hildebrandt F (2011) Exome sequencing reveals
Kaito H, Iijima K, Ito S (2014) Rituximab treatment combined cubilin mutation as a single-gene cause of proteinuria. J Am Soc
with methylprednisolone pulse therapy and immunosuppressants Nephrol 22:1815–1820
for childhood steroid-resistant nephrotic syndrome. Pediatr 118. Amsellem S, Gburek J, Hamard G, Nielsen R, Willnow TE,
Nephrol 29:1181–1187 Devuyst O, Nexo E, Verroust PJ, Christensen EI, Kozyraki R
102. Fornoni A, Sageshima J, Wei C, Merscher-Gomez S, Aguillon- (2010) Cubilin is essential for albumin reabsorption in the renal
Prada R, Jauregui AN, Li J, Mattiazzi A, Ciancio G, Chen L, proximal tubule. J Am Soc Nephrol 21:1859–1867
Zilleruelo G, Abitbol C, Chandar J, Seeherunvong W, Ricordi C, 119. Winn MP, Conlon PJ, Lynn KL, Farrington MK, Creazzo T,
Ikehata M, Rastaldi MP, Reiser J, Burke GW 3rd (2011) Hawkins AF, Daskalakis N, Kwan SY, Ebersviller S, Burchette
Rituximab targets podocytes in recurrent focal segmental JL, Pericak-Vance MA, Howell DN, Vance JM, Rosenberg PB
glomerulosclerosis. Sci Transl Med 3:85ra46 (2005) A mutation in the TRPC6 cation channel causes familial
103. Hall G, Gbadegesin RA (2015) Translating genetic findings in focal segmental glomerulosclerosis. Science 308:1801–1804
hereditary nephrotic syndrome: the missing loops. Am J Physiol 120. Urban N, Hill K, Wang L, Kuebler WM, Schaefer M (2012) Novel
Renal Physiol 309:F24–F28 pharmacological TRPC inhibitors block hypoxia-induced vaso-
104. Turunen M, Olsson J, Dallner G (2004) Metabolism and function constriction. Cell Calcium 51:194–206
of coenzyme Q. Biochim Biophys Acta 1660:171–199 121. Zhou Y, Castonguay P, Sidhom EH, Clark AR, Dvela-Levitt M,
105. Tran UC, Clarke CF (2007) Endogenous synthesis of coenzyme Q Kim S, Sieber J, Wieder N, Jung JY, Andreeva S, Reichardt J,
in eukaryotes. Mitochondrion (7 Suppl):S62–S71 Dubois F, Hoffmann SC, Basgen JM, Montesinos MS, Weins A,
106. Crane FL (2007) Discovery of ubiquinone (coenzyme Q) and an Johnson AC, Lander ES, Garrett MR, Hopkins CR, Greka A
overview of function. Mitochondrion (7 Suppl):S2–S7 (2017) A small-molecule inhibitor of TRPC5 ion channels sup-
107. Bentinger M, Brismar K, Dallner G (2007) The antioxidant role of presses progressive kidney disease in animal models. Science 358:
coenzyme Q. Mitochondrion (7 Suppl):S41–S50 1332–1336
108. Doimo M, Desbats MA, Cerqua C, Cassina M, Trevisson E, 122. Hall GLB, Khan K, Pediaditakis I, Xiao J, Wu G, Wang L,
Salviati L (2014) Genetics of coenzyme q10 deficiency. Mol Kovalik ME, Chryst-Stangl M, Davis EE, Spurney RF,
Syndromol 5:156–162 Gbadegesin RA (2018) The human FSGS-causing ANLN
Pediatr Nephrol

R431C mutation induces dysregulated PI3K/AKT/mTOR/Rac1 127. Pelletier JHKK, Engen R, Bensimhon A, Varner J, Rheault M,
signaling in podocytes. J Am Soc Nephrol 29:2011–2122 Srivastava T, Straatmann C, Silva C, Davis TK, Wenderfer S,
123. Bagga A, Mudigoudar BD, Hari P, Vasudev V (2004) Enalapril Gibson K, Selewski D, Barcia J, Weng P, Licht C, Jawa N,
dosage in steroid-resistant nephrotic syndrome. Pediatr Nephrol Kallash M, Foreman JW, Wigfall DR, Chua AN, Chambers E,
19:45–50 Hornik CP, Brewer ED, Nagaraj SK, Greenbaum LA,
124. Li Z, Duan C, He J, Wu T, Xun M, Zhang Y, Yin Y (2010) Gbadegesin RA (2018) Recurrence of nephrotic syndrome follow-
Mycophenolate mofetil therapy for children with steroid- ing kidney transplantation is associated with initial native kidney
resistant nephrotic syndrome. Pediatr Nephrol 25:883–888 biopsy findings. Pediatr Nephrol 33:1773–1780
125. Wada T, Nangaku M (2015) A circulating permeability factor in 128. Winn MP, Alkhunaizi AM, Bennett WM, Garber RL, Howell DN,
focal segmental glomerulosclerosis: the hunt continues. Clin B u t t e r l y D W, C o n l o n P J ( 1 9 9 9 ) F o c a l s e g m e n t a l
Kidney J 8:708–715 glomerulosclerosis: a need for caution in live-related renal trans-
126. Konigshausen E, Sellin L (2016) Circulating permeability factors plantation. Am J Kidney Dis 33:970–974
in primary focal segmental glomerulosclerosis: a review of pro-
posed candidates. Biomed Res Int 2016:3765608

You might also like